Multi-Omics Analysis Reveals the transforming growth factor-β Signaling-Driven Multicellular Interactions with Prognostic Relevance in Cervical Cancer Progression.

IF 3.3 3区 医学 Q2 ONCOLOGY
Journal of Cancer Pub Date : 2025-06-20 eCollection Date: 2025-01-01 DOI:10.7150/jca.114505
Yuhan Wang, Guangxu Cao, Huimin Zeng, Yong Zhi, Mengting Xu, Ying Wang, Min Liu, Yetian Ruan, Ka Yu Tse, Qingfeng Zhang, Jinli Gao, Zhiqiang Han, Fang Li
{"title":"Multi-Omics Analysis Reveals the transforming growth factor-β Signaling-Driven Multicellular Interactions with Prognostic Relevance in Cervical Cancer Progression.","authors":"Yuhan Wang, Guangxu Cao, Huimin Zeng, Yong Zhi, Mengting Xu, Ying Wang, Min Liu, Yetian Ruan, Ka Yu Tse, Qingfeng Zhang, Jinli Gao, Zhiqiang Han, Fang Li","doi":"10.7150/jca.114505","DOIUrl":null,"url":null,"abstract":"<p><p>While cervical cancer (CC) prognosis depends on tumor staging, the spatiotemporal evolution of tumor microenvironment (TME) heterogeneity during metastatic progression remains poorly characterized at single-cell resolution. We employed an integrative multi-omics approach, combining single-cell RNA sequencing (scRNA-seq; n = 11), spatial transcriptomics (ST), and bulk RNA-seq data from the TCGA-CESC cohort (n = 304), to systematically map TME remodeling across CC progression stages. scRNA-seq was performed on primary lesions from patients with localized (n = 3), regional (n = 4), and metastatic (n = 4) diseases, with in-depth analyses focusing on cellular characteristics, cell type composition alterations, functional changes, differentiation trajectories, and cell-cell interaction networks. These findings were further validated using spatial transcriptomics, bulk RNA-seq data, and multiple immunohistochemistry (mIHC) experiments. ScRNA-seq data revealed that the TME of the metastatic group displayed a distinct immunosuppressive phenotype. Three key subclusters closely linked to TME remodeling in this group were identified. Notably, a novel metastasis-associated epithelial subpopulation (Epi0_AGR2), characterized by both epithelial-mesenchymal transition (EMT) and chemokine secretory phenotypes, was discovered. Gene Set Variation Analysis (GSVA) revealed that transforming growth factor β (TGF-β) signaling activation served as its primary transcriptional driver. Additionally, a neutrophil subset with pro-tumor and immunosuppressive properties, as well as a cancer-associated fibroblasts (CAFs) subset that promoted angiogenesis, were enriched in the metastatic group. Cell-cell interaction analysis and spatial mapping further revealed the formation of coordinated Epi0-neutrophil-CAFs niches, which established TGF-β-CXCL1/2/8-OSM/OSMR feedforward loops. Importantly, a computational model derived from the TME metastatic niche signature demonstrated significant prognostic stratification in the TCGA cohort (HR = 2.5179, p = 0.0144). In all, this study provides the first comprehensive delineation of stage-specific TME dynamics in CC, revealing TGF-β-driven cellular cooperativity as a metastatic switch. The joint framework establishes a potential clinically translatable tool for precision prognosis and therapeutic targeting.</p>","PeriodicalId":15183,"journal":{"name":"Journal of Cancer","volume":"16 9","pages":"2857-2876"},"PeriodicalIF":3.3000,"publicationDate":"2025-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12244011/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/jca.114505","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2025/1/1 0:00:00","PubModel":"eCollection","JCR":"Q2","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

While cervical cancer (CC) prognosis depends on tumor staging, the spatiotemporal evolution of tumor microenvironment (TME) heterogeneity during metastatic progression remains poorly characterized at single-cell resolution. We employed an integrative multi-omics approach, combining single-cell RNA sequencing (scRNA-seq; n = 11), spatial transcriptomics (ST), and bulk RNA-seq data from the TCGA-CESC cohort (n = 304), to systematically map TME remodeling across CC progression stages. scRNA-seq was performed on primary lesions from patients with localized (n = 3), regional (n = 4), and metastatic (n = 4) diseases, with in-depth analyses focusing on cellular characteristics, cell type composition alterations, functional changes, differentiation trajectories, and cell-cell interaction networks. These findings were further validated using spatial transcriptomics, bulk RNA-seq data, and multiple immunohistochemistry (mIHC) experiments. ScRNA-seq data revealed that the TME of the metastatic group displayed a distinct immunosuppressive phenotype. Three key subclusters closely linked to TME remodeling in this group were identified. Notably, a novel metastasis-associated epithelial subpopulation (Epi0_AGR2), characterized by both epithelial-mesenchymal transition (EMT) and chemokine secretory phenotypes, was discovered. Gene Set Variation Analysis (GSVA) revealed that transforming growth factor β (TGF-β) signaling activation served as its primary transcriptional driver. Additionally, a neutrophil subset with pro-tumor and immunosuppressive properties, as well as a cancer-associated fibroblasts (CAFs) subset that promoted angiogenesis, were enriched in the metastatic group. Cell-cell interaction analysis and spatial mapping further revealed the formation of coordinated Epi0-neutrophil-CAFs niches, which established TGF-β-CXCL1/2/8-OSM/OSMR feedforward loops. Importantly, a computational model derived from the TME metastatic niche signature demonstrated significant prognostic stratification in the TCGA cohort (HR = 2.5179, p = 0.0144). In all, this study provides the first comprehensive delineation of stage-specific TME dynamics in CC, revealing TGF-β-driven cellular cooperativity as a metastatic switch. The joint framework establishes a potential clinically translatable tool for precision prognosis and therapeutic targeting.

多组学分析揭示了转化生长因子-β信号驱动的多细胞相互作用与宫颈癌进展的预后相关性。
虽然宫颈癌(CC)的预后取决于肿瘤分期,但在转移进展过程中肿瘤微环境(TME)异质性的时空演变在单细胞分辨率上仍然很差。我们采用了综合多组学方法,结合单细胞RNA测序(scRNA-seq;n = 11),来自TCGA-CESC队列(n = 304)的空间转录组学(ST)和大量RNA-seq数据,系统地绘制CC进展阶段的TME重塑。scRNA-seq对局限性(n = 3)、区域性(n = 4)和转移性(n = 4)疾病患者的原发病变进行了深入分析,重点是细胞特征、细胞类型组成改变、功能改变、分化轨迹和细胞-细胞相互作用网络。这些发现通过空间转录组学、大量RNA-seq数据和多次免疫组织化学(mIHC)实验得到了进一步验证。ScRNA-seq数据显示,转移组的TME表现出明显的免疫抑制表型。在这一组中确定了三个与TME重塑密切相关的关键亚群。值得注意的是,研究人员发现了一种新的转移相关上皮亚群(Epi0_AGR2),其特征是上皮-间质转化(EMT)和趋化因子分泌表型。基因集变异分析(GSVA)显示,转化生长因子β (TGF-β)信号激活是其主要的转录驱动因子。此外,具有促肿瘤和免疫抑制特性的中性粒细胞亚群,以及促进血管生成的癌症相关成纤维细胞(CAFs)亚群,在转移组中富集。细胞-细胞相互作用分析和空间作图进一步揭示了epi0 - neutrophill - cafs协同生态位的形成,并建立了TGF-β-CXCL1/2/8-OSM/OSMR前反馈回路。重要的是,基于TME转移生态位特征的计算模型显示了TCGA队列中显著的预后分层(HR = 2.5179, p = 0.0144)。总之,本研究首次全面描述了肝癌分期特异性TME动力学,揭示了TGF-β驱动的细胞协同性作为转移开关。联合框架建立了一个潜在的临床翻译工具,用于精确预后和治疗靶向。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Journal of Cancer
Journal of Cancer ONCOLOGY-
CiteScore
8.10
自引率
2.60%
发文量
333
审稿时长
12 weeks
期刊介绍: Journal of Cancer is an open access, peer-reviewed journal with broad scope covering all areas of cancer research, especially novel concepts, new methods, new regimens, new therapeutic agents, and alternative approaches for early detection and intervention of cancer. The Journal is supported by an international editorial board consisting of a distinguished team of cancer researchers. Journal of Cancer aims at rapid publication of high quality results in cancer research while maintaining rigorous peer-review process.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信